Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Cell ; 71(2): 343-351.e4, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30029007

ABSTRACT

Class II phosphoinositide 3-kinases (PI3K-C2) are large multidomain enzymes that control cellular functions ranging from membrane dynamics to cell signaling via synthesis of 3'-phosphorylated phosphoinositides. Activity of the alpha isoform (PI3K-C2α) is associated with endocytosis, angiogenesis, and glucose metabolism. How PI3K-C2α activity is controlled at sites of endocytosis remains largely enigmatic. Here we show that the lipid-binding PX-C2 module unique to class II PI3Ks autoinhibits kinase activity in solution but is essential for full enzymatic activity at PtdIns(4,5)P2-rich membranes. Using HDX-MS, we show that the PX-C2 module folds back onto the kinase domain, inhibiting its basal activity. Destabilization of this intramolecular contact increases PI3K-C2α activity in vitro and in cells, leading to accumulation of its lipid product, increased recruitment of the endocytic effector SNX9, and facilitated endocytosis. Our studies uncover a regulatory mechanism in which coincident binding of phosphoinositide substrate and cofactor selectively activate PI3K-C2α at sites of endocytosis.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/metabolism , Class II Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , C2 Domains/physiology , COS Cells , Chlorocebus aethiops , Class I Phosphatidylinositol 3-Kinases/metabolism , Class I Phosphatidylinositol 3-Kinases/physiology , Clathrin/physiology , Endocytosis/physiology , HEK293 Cells , Homeostasis , Humans , Lipids/physiology , Mass Spectrometry , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Protein Domains , Signal Transduction
2.
Gut ; 72(2): 360-371, 2023 02.
Article in English | MEDLINE | ID: mdl-35623884

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with limited therapeutic options. However, metabolic adaptation to the harsh PDAC environment can expose liabilities useful for therapy. Targeting the key metabolic regulator mechanistic target of rapamycin complex 1 (mTORC1) and its downstream pathway shows efficacy only in subsets of patients but gene modifiers maximising response remain to be identified. DESIGN: Three independent cohorts of PDAC patients were studied to correlate PI3K-C2γ protein abundance with disease outcome. Mechanisms were then studied in mouse (KPC mice) and cellular models of PDAC, in presence or absence of PI3K-C2γ (WT or KO). PI3K-C2γ-dependent metabolic rewiring and its impact on mTORC1 regulation were assessed in conditions of limiting glutamine availability. Finally, effects of a combination therapy targeting mTORC1 and glutamine metabolism were studied in WT and KO PDAC cells and preclinical models. RESULTS: PI3K-C2γ expression was reduced in about 30% of PDAC cases and was associated with an aggressive phenotype. Similarly, loss of PI3K-C2γ in KPC mice enhanced tumour development and progression. The increased aggressiveness of tumours lacking PI3K-C2γ correlated with hyperactivation of mTORC1 pathway and glutamine metabolism rewiring to support lipid synthesis. PI3K-C2γ-KO tumours failed to adapt to metabolic stress induced by glutamine depletion, resulting in cell death. CONCLUSION: Loss of PI3K-C2γ prevents mTOR inactivation and triggers tumour vulnerability to RAD001 (mTOR inhibitor) and BPTES/CB-839 (glutaminase inhibitors). Therefore, these results might open the way to personalised treatments in PDAC with PI3K-C2γ loss.


Subject(s)
Carcinoma, Pancreatic Ductal , Everolimus , Lipids , Lysosomes , MTOR Inhibitors , Pancreatic Neoplasms , Phosphatidylinositol 3-Kinases , Animals , Mice , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Proliferation , Glutamine/metabolism , Lipids/biosynthesis , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Nutrients , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Everolimus/therapeutic use , MTOR Inhibitors/therapeutic use , Glutaminase , Pancreatic Neoplasms
3.
PLoS Genet ; 15(4): e1008088, 2019 04.
Article in English | MEDLINE | ID: mdl-31034465

ABSTRACT

PIK3C2A is a class II member of the phosphoinositide 3-kinase (PI3K) family that catalyzes the phosphorylation of phosphatidylinositol (PI) into PI(3)P and the phosphorylation of PI(4)P into PI(3,4)P2. At the cellular level, PIK3C2A is critical for the formation of cilia and for receptor mediated endocytosis, among other biological functions. We identified homozygous loss-of-function mutations in PIK3C2A in children from three independent consanguineous families with short stature, coarse facial features, cataracts with secondary glaucoma, multiple skeletal abnormalities, neurological manifestations, among other findings. Cellular studies of patient-derived fibroblasts found that they lacked PIK3C2A protein, had impaired cilia formation and function, and demonstrated reduced proliferative capacity. Collectively, the genetic and molecular data implicate mutations in PIK3C2A in a new Mendelian disorder of PI metabolism, thereby shedding light on the critical role of a class II PI3K in growth, vision, skeletal formation and neurological development. In particular, the considerable phenotypic overlap, yet distinct features, between this syndrome and Lowe's syndrome, which is caused by mutations in the PI-5-phosphatase OCRL, highlight the key role of PI metabolizing enzymes in specific developmental processes and demonstrate the unique non-redundant functions of each enzyme. This discovery expands what is known about disorders of PI metabolism and helps unravel the role of PIK3C2A and class II PI3Ks in health and disease.


Subject(s)
Bone Diseases, Developmental/genetics , Cataract/genetics , Ciliary Motility Disorders/genetics , Dwarfism/genetics , Mutation , Phosphatidylinositol 3-Kinases/genetics , Adolescent , Adult , Child , Consanguinity , Female , Fibroblasts/metabolism , Humans , Male , Pedigree , Phenotype , Young Adult
4.
Nature ; 499(7457): 233-7, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23823722

ABSTRACT

Phosphoinositides serve crucial roles in cell physiology, ranging from cell signalling to membrane traffic. Among the seven eukaryotic phosphoinositides the best studied species is phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2), which is concentrated at the plasma membrane where, among other functions, it is required for the nucleation of endocytic clathrin-coated pits. No phosphatidylinositol other than PI(4,5)P2 has been implicated in clathrin-mediated endocytosis, whereas the subsequent endosomal stages of the endocytic pathway are dominated by phosphatidylinositol-3-phosphates(PI(3)P). How phosphatidylinositol conversion from PI(4,5)P2-positive endocytic intermediates to PI(3)P-containing endosomes is achieved is unclear. Here we show that formation of phosphatidylinositol-3,4-bisphosphate (PI(3,4)P2) by class II phosphatidylinositol-3-kinase C2α (PI(3)K C2α) spatiotemporally controls clathrin-mediated endocytosis. Depletion of PI(3,4)P2 or PI(3)K C2α impairs the maturation of late-stage clathrin-coated pits before fission. Timed formation of PI(3,4)P2 by PI(3)K C2α is required for selective enrichment of the BAR domain protein SNX9 at late-stage endocytic intermediates. These findings provide a mechanistic framework for the role of PI(3,4)P2 in endocytosis and unravel a novel discrete function of PI(3,4)P2 in a central cell physiological process.


Subject(s)
Endocytosis , Phosphatidylinositol Phosphates/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Class II Phosphatidylinositol 3-Kinases/metabolism , Clathrin-Coated Vesicles/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Phosphoric Monoester Hydrolases/metabolism , Sorting Nexins/metabolism , Time Factors
5.
J Cell Biochem ; 118(11): 3561-3568, 2017 11.
Article in English | MEDLINE | ID: mdl-28419521

ABSTRACT

Cytokinetic abscission involves the fine and regulated recruitment of membrane remodeling proteins that participate in the abscission of the intracellular bridge that connects the two dividing cells. This essential process is mediated by the concomitant activity of the endosomal sorting complex required for transport (ESCRT) and the vesicular trafficking directed to the midbody. Phosphoinositides (PtdIns), produced at plasma membrane, and endosomes, act as molecular intermediates by recruiting effector proteins involved in multiple cellular processes, such as intracellular signaling, endo- and exo-cytosis, and membrane remodeling events. Emerging evidences suggest that PtdIns have an active role in recruiting key elements that control the stability and the remodeling of the cytoskeleton from the furrow ingression to the abscission, at the end of cytokinesis. Accordingly, a possible concomitant and coordinated activity between PtdIns production and ESCRT machinery assembly could also exist and recent findings are pointing the attention on poorly understood ESCRT subunits potentially able to associate with PtdIns rich membranes. Although further studies are required to link PtdIns to ESCRT machinery during abscission, this might represent a promising field of study. J. Cell. Biochem. 118: 3561-3568, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Cell Membrane/metabolism , Cytokinesis/physiology , Endosomal Sorting Complexes Required for Transport/metabolism , Phosphatidylinositols/metabolism , Animals , Humans
6.
J Am Soc Nephrol ; 27(4): 1135-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26271513

ABSTRACT

Signaling from the primary cilium regulates kidney tubule development and cyst formation. However, the mechanism controlling targeting of ciliary components necessary for cilium morphogenesis and signaling is largely unknown. Here, we studied the function of class II phosphoinositide 3-kinase-C2α (PI3K-C2α) in renal tubule-derived inner medullary collecting duct 3 cells and show that PI3K-C2α resides at the recycling endosome compartment in proximity to the primary cilium base. In this subcellular location, PI3K-C2α controlled the activation of Rab8, a key mediator of cargo protein targeting to the primary cilium. Consistently, partial reduction of PI3K-C2α was sufficient to impair elongation of the cilium and the ciliary transport of polycystin-2, as well as to alter proliferation signals linked to polycystin activity. In agreement, heterozygous deletion of PI3K-C2α in mice induced cilium elongation defects in kidney tubules and predisposed animals to cyst development, either in genetic models of polycystin-1/2 reduction or in response to ischemia/reperfusion-induced renal damage. These results indicate that PI3K-C2α is required for the transport of ciliary components such as polycystin-2, and partial loss of this enzyme is sufficient to exacerbate the pathogenesis of cystic kidney disease.


Subject(s)
Cilia/physiology , Class II Phosphatidylinositol 3-Kinases/physiology , Kidney Diseases, Cystic , TRPP Cation Channels/physiology , Animals , Kidney Diseases, Cystic/etiology , Male , Mice , Signal Transduction
7.
Cell Rep ; 43(6): 114273, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38843397

ABSTRACT

Phosphoinositides (PtdIns) are a family of differentially phosphorylated lipid second messengers localized to the cytoplasmic leaflet of both plasma and intracellular membranes. Kinases and phosphatases can selectively modify the PtdIns composition of different cellular compartments, leading to the recruitment of specific binding proteins, which control cellular homeostasis and proliferation. Thus, while PtdIns affect cell growth and survival during interphase, they are also emerging as key drivers in multiple temporally defined membrane remodeling events of mitosis, like cell rounding, spindle orientation, cytokinesis, and abscission. In this review, we summarize and discuss what is known about PtdIns function during mitosis and how alterations in the production and removal of PtdIns can interfere with proper cell division.


Subject(s)
Mitosis , Phosphatidylinositols , Humans , Phosphatidylinositols/metabolism , Animals , Cytokinesis/physiology
8.
Sci Signal ; 17(838): eadp3504, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38805585

ABSTRACT

The Hippo pathway blocks epithelial-mesenchymal transition and metastasis in cancer mediated by the transcriptional coactivator YAP. In this issue of Science Signaling, Palamiuc et al. demonstrate that phosphatidylinositol 5-phosphate (PI5P) enhances Hippo pathway activation and that simultaneously the Hippo pathway initiates a positive feedback loop by inhibiting the conversion of PI5P into PIP2.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Phosphatidylinositol Phosphates , Protein Serine-Threonine Kinases , Signal Transduction , Humans , Phosphatidylinositol Phosphates/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , Animals , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Hippo Signaling Pathway , Transcription Factors/metabolism , Transcription Factors/genetics
9.
Nat Struct Mol Biol ; 29(3): 218-228, 2022 03.
Article in English | MEDLINE | ID: mdl-35256802

ABSTRACT

Phosphatidylinositol 3-kinase type 2α (PI3KC2α) is an essential member of the structurally unresolved class II PI3K family with crucial functions in lipid signaling, endocytosis, angiogenesis, viral replication, platelet formation and a role in mitosis. The molecular basis of these activities of PI3KC2α is poorly understood. Here, we report high-resolution crystal structures as well as a 4.4-Å cryogenic-electron microscopic (cryo-EM) structure of PI3KC2α in active and inactive conformations. We unravel a coincident mechanism of lipid-induced activation of PI3KC2α at membranes that involves large-scale repositioning of its Ras-binding and lipid-binding distal Phox-homology and C-C2 domains, and can serve as a model for the entire class II PI3K family. Moreover, we describe a PI3KC2α-specific helical bundle domain that underlies its scaffolding function at the mitotic spindle. Our results advance our understanding of PI3K biology and pave the way for the development of specific inhibitors of class II PI3K function with wide applications in biomedicine.


Subject(s)
Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases , Endocytosis , Lipids , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
10.
Adv Sci (Weinh) ; 9(9): e2103249, 2022 03.
Article in English | MEDLINE | ID: mdl-35098698

ABSTRACT

Breast cancer is the most prevalent cancer and a major cause of death in women worldwide. Although early diagnosis and therapeutic intervention significantly improve patient survival rate, metastasis still accounts for most deaths. Here it is reported that, in a cohort of more than 2000 patients with breast cancer, overexpression of PI3KC2α occurs in 52% of cases and correlates with high tumor grade as well as increased probability of distant metastatic events, irrespective of the subtype. Mechanistically, it is demonstrated that PI3KC2α synthetizes a pool of PI(3,4)P2 at focal adhesions that lowers their stability and directs breast cancer cell migration, invasion, and metastasis. PI(3,4)P2 locally produced by PI3KC2α at focal adhesions recruits the Ras GTPase activating protein 3 (RASA3), which inactivates R-RAS, leading to increased focal adhesion turnover, migration, and invasion both in vitro and in vivo. Proof-of-concept is eventually provided that inhibiting PI3KC2α or lowering RASA3 activity at focal adhesions significantly reduces the metastatic burden in PI3KC2α-overexpressing breast cancer, thereby suggesting a novel strategy for anti-breast cancer therapy.


Subject(s)
Breast Neoplasms , Cell Adhesion/physiology , Female , Focal Adhesions/metabolism , Focal Adhesions/pathology , GTPase-Activating Proteins/metabolism , Humans , Phosphatidylinositols/metabolism
11.
Cancers (Basel) ; 13(14)2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34298731

ABSTRACT

Breast cancer is the most frequently diagnosed cancer and the primary cause of cancer death in women worldwide. Although early diagnosis and cancer growth inhibition has significantly improved breast cancer survival rate over the years, there is a current need to develop more effective systemic treatments to prevent metastasis. One of the most commonly altered pathways driving breast cancer cell growth, survival, and motility is the PI3K/AKT/mTOR signaling cascade. In the past 30 years, a great surge of inhibitors targeting these key players has been developed at a rapid pace, leading to effective preclinical studies for cancer therapeutics. However, the central role of PI3K/AKT/mTOR signaling varies among diverse biological processes, suggesting the need for more specific and sophisticated strategies for their use in cancer therapy. In this review, we provide a perspective on the role of the PI3K signaling pathway and the most recently developed PI3K-targeting breast cancer therapies.

12.
Science ; 374(6573): eabk0410, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34882480

ABSTRACT

Cytokinetic membrane abscission is a spatially and temporally regulated process that requires ESCRT (endosomal sorting complexes required for transport)­dependent control of membrane remodeling at the midbody, a subcellular organelle that defines the cleavage site. Alteration of ESCRT function can lead to cataract, but the underlying mechanism and its relation to cytokinesis are unclear. We found a lens-specific cytokinetic process that required PI3K-C2α (phosphatidylinositol-4-phosphate 3-kinase catalytic subunit type 2α), its lipid product PI(3,4)P2 (phosphatidylinositol 3,4-bisphosphate), and the PI(3,4)P2­binding ESCRT-II subunit VPS36 (vacuolar protein-sorting-associated protein 36). Loss of each of these components led to impaired cytokinesis, triggering premature senescence in the lens of fish, mice, and humans. Thus, an evolutionarily conserved pathway underlies the cell type­specific control of cytokinesis that helps to prevent early onset cataract by protecting from senescence.


Subject(s)
Cataract/pathology , Cellular Senescence , Cytokinesis , Endosomal Sorting Complexes Required for Transport/metabolism , Lens, Crystalline/cytology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Aging, Premature , Animals , Biological Evolution , Calcium-Binding Proteins/metabolism , Cataract/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Humans , Lens, Crystalline/growth & development , Lens, Crystalline/metabolism , Mice , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Tubulin/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Adv Biol Regul ; 75: 100693, 2020 01.
Article in English | MEDLINE | ID: mdl-32008962

ABSTRACT

Phosphoinositides (PI) are key players in many trafficking and signaling pathways. Recent advances regarding the synthesis, location and functions of these lipids have improved our understanding of how and when these lipids are generated and what their roles are in physiology and disease. In particular, PI play a central role in the regulation of cell proliferation and metabolism. Here, we will review recent advances in our understanding of PI function, regulation, and importance in different aspects of proliferation and energy metabolism.


Subject(s)
Energy Metabolism , Phosphatidylinositols/metabolism , Signal Transduction , Animals , Humans , Phosphatidylinositols/genetics , Protein Transport
14.
Front Oncol ; 10: 360, 2020.
Article in English | MEDLINE | ID: mdl-32296634

ABSTRACT

The phosphatidylinositide 3 kinases (PI3Ks) and their downstream mediators AKT and mammalian target of rapamycin (mTOR) are central regulators of glycolysis, cancer metabolism, and cancer cell proliferation. At the molecular level, PI3K signaling involves the generation of the second messenger lipids phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] and phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2]. There is increasing evidence that PI(3,4)P2 is not only the waste product for the removal of PI(3,4,5)P3 but can also act as a signaling molecule. The selective cellular functions for PI(3,4)P2 independent of PI(3,4,5)P3 have been recently described, including clathrin-mediated endocytosis and mTOR regulation. However, the specific spatiotemporal dynamics and signaling role of PI3K minor lipid messenger PI(3,4)P2 are not well-understood. This review aims at highlighting the biological functions of this lipid downstream of phosphoinositide kinases and phosphatases and its implication in cancer metabolism.

15.
Biochim Biophys Acta Rev Cancer ; 1871(2): 361-366, 2019 04.
Article in English | MEDLINE | ID: mdl-30946868

ABSTRACT

The key role of phosphoinositide 3-kinase (PI3K) pathway in different cellular processes and several disorders, together with the presence of targetable proteins, opened the way to promising studies for the development of small molecule inhibitors. Despite the high expectation, the shift of PI3K inhibitors to the clinic met several limitations due to the emergence of dose-limiting, on-target adverse effects. In this review, we will summarize the main issues and recent advances in PI3K inhibitors clinical trials. The effort to develop isoform-specific inhibitors, together with novel therapeutic strategies aimed at reducing the toxicity and adverse effects, opened a new promising era for PI3K inhibitors. In addition, we will focus on the recent emergence of class II and III PI3K inhibitors, which helped to define their class I non-redundant role.


Subject(s)
Neoplasms/enzymology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/physiology , Animals , Antineoplastic Agents/pharmacology , Humans , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Neoplasms/drug therapy
16.
Trends Cell Biol ; 29(4): 339-359, 2019 04.
Article in English | MEDLINE | ID: mdl-30691999

ABSTRACT

The phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that phosphorylate inositol phospholipids, thereby controlling membrane lipid composition and regulating a wide range of intracellular processes, including vesicular trafficking and signal transduction. Despite the vast knowledge on class I PI3Ks, recent studies are only now revealing the importance of class II PI3Ks in cell proliferation, survival, and migration. Increasing evidence suggests that the three class II PI3Ks isoforms (PI3K-C2α, PI3K-C2ß, and PI3K-C2γ) have distinct and non-overlapping cellular roles. Here, we focus on the cellular functions of class II PI3Ks in different cell systems and underline the emerging importance of these enzymes in various physiological and pathological contexts.


Subject(s)
Cells/cytology , Cells/enzymology , Class II Phosphatidylinositol 3-Kinases/metabolism , Metabolic Diseases/enzymology , Neoplasms/enzymology , Animals , Cells/pathology , Humans , Metabolic Diseases/pathology , Neoplasms/pathology , Signal Transduction
17.
J Exp Clin Cancer Res ; 38(1): 472, 2019 Nov 21.
Article in English | MEDLINE | ID: mdl-31752944

ABSTRACT

BACKGROUND: Alteration of signalling pathways regulating cell cycle progression is a common feature of cancer cells. Several drugs targeting distinct phases of the cell cycle have been developed but the inability of many of them to discriminate between normal and cancer cells has strongly limited their clinical potential because of their reduced efficacy at the concentrations used to limit adverse side effects. Mechanisms of resistance have also been described, further affecting their efficacy. Identification of novel targets that can potentiate the effect of these drugs or overcome drug resistance can provide a useful strategy to exploit the anti-cancer properties of these agents to their fullest. METHODS: The class II PI3K isoform PI3K-C2ß was downregulated in prostate cancer PC3 cells and cervical cancer HeLa cells using selective siRNAs and the effect on cell growth was determined in the absence or presence of the microtubule-stabilizing agent/anti-cancer drug docetaxel. Mitosis progression was monitored by time-lapse microscopy. Clonogenic assays were performed to determine the ability of PC3 and HeLa cells to form colonies upon PI3K-C2ß downregulation in the absence or presence of docetaxel. Cell multi-nucleation was assessed by immunofluorescence. Tumour growth in vivo was assessed using a xenograft model of PC3 cells upon PI3K-C2ß downregulation and in combination with docetaxel. RESULTS: Downregulation of PI3K-C2ß delays mitosis progression in PC3 and HeLa cells, resulting in reduced ability to form colonies in clonogenic assays in vitro. Compared to control cells, PC3 cells lacking PI3K-C2ß form smaller and more compact colonies in vitro and they form tumours more slowly in vivo in the first weeks after cells implant. Stable and transient PI3K-C2ß downregulation potentiates the effect of low concentrations of docetaxel on cancer cell growth. Combination of PI3K-C2ß downregulation and docetaxel almost completely prevents colonies formation in clonogenic assays in vitro and strongly inhibits tumour growth in vivo. CONCLUSIONS: These data reveal a novel role for the class II PI3K PI3K-C2ß during mitosis progression. Furthermore, data indicate that blockade of PI3K-C2ß might represent a novel strategy to potentiate the effect of docetaxel on cancer cell growth.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/metabolism , Docetaxel/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/enzymology , Animals , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Proliferation/drug effects , Down-Regulation , Female , HeLa Cells , Humans , Male , Mice, Nude , PC-3 Cells , Prostatic Neoplasms/pathology , Random Allocation , Transfection , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Cancer Cell ; 32(4): 444-459.e7, 2017 10 09.
Article in English | MEDLINE | ID: mdl-29017056

ABSTRACT

Proper organization of the mitotic spindle is key to genetic stability, but molecular components of inter-microtubule bridges that crosslink kinetochore fibers (K-fibers) are still largely unknown. Here we identify a kinase-independent function of class II phosphoinositide 3-OH kinase α (PI3K-C2α) acting as limiting scaffold protein organizing clathrin and TACC3 complex crosslinking K-fibers. Downregulation of PI3K-C2α causes spindle alterations, delayed anaphase onset, and aneuploidy, indicating that PI3K-C2α expression is required for genomic stability. Reduced abundance of PI3K-C2α in breast cancer models initially impairs tumor growth but later leads to the convergent evolution of fast-growing clones with mitotic checkpoint defects. As a consequence of altered spindle, loss of PI3K-C2α increases sensitivity to taxane-based therapy in pre-clinical models and in neoadjuvant settings.


Subject(s)
Breast Neoplasms/pathology , Genomic Instability , Phosphatidylinositol 3-Kinases/physiology , Spindle Apparatus/physiology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Cycle Proteins/physiology , Cell Proliferation , Humans , MCF-7 Cells , Mad2 Proteins/physiology , Mice , Microtubule-Associated Proteins/physiology , Nuclear Proteins/physiology , Taxoids/therapeutic use
20.
Cancer Discov ; 5(7): 740-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25883022

ABSTRACT

UNLABELLED: The phosphatases PTEN and INPP4B have been proposed to act as tumor suppressors by antagonizing PI3K-AKT signaling and are frequently dysregulated in human cancer. Although PTEN has been extensively studied, little is known about the underlying mechanisms by which INPP4B exerts its tumor-suppressive function and its role in tumorigenesis in vivo. Here, we show that a partial or complete loss of Inpp4b morphs benign thyroid adenoma lesions in Pten heterozygous mice into lethal and metastatic follicular-like thyroid cancer (FTC). Importantly, analyses of human thyroid cancer cell lines and specimens reveal INPP4B downregulation in FTC. Mechanistically, we find that INPP4B, but not PTEN, is enriched in the early endosomes of thyroid cancer cells, where it selectively inhibits AKT2 activation and in turn tumor proliferation and anchorage-independent growth. We therefore identify INPP4B as a novel tumor suppressor in FTC oncogenesis and metastasis through localized regulation of the PI3K-AKT pathway at the endosomes. SIGNIFICANCE: Although both PTEN and INPP4B can inhibit PI3K-AKT signaling through their lipid phosphatase activities, here we demonstrate lack of an epistatic relationship between the two tumor suppressors. Instead, the qualitative regulation of PI3K-AKT2 signaling by INPP4B provides a mechanism for their cooperation in suppressing thyroid tumorigenesis and metastasis.


Subject(s)
Adenocarcinoma, Follicular/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thyroid Neoplasms/pathology , Adenocarcinoma, Follicular/genetics , Animals , Cell Line, Tumor , Endosomes/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Mice , Neoplasm Metastasis , PTEN Phosphohydrolase/metabolism , Phosphoric Monoester Hydrolases/genetics , Signal Transduction , Thyroid Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL